Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in...

13
Virulence-Associated Enzymes of Cryptococcus neoformans Fausto Almeida, a,b Julie M. Wolf, a Arturo Casadevall a,c Department of Microbiology and Immunology, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, USA a ; Department of Cellular and Molecular Biology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazil b ; Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA c Enzymes play key roles in fungal pathogenesis. Manipulation of enzyme expression or activity can significantly alter the infec- tion process, and enzyme expression profiles can be a hallmark of disease. Hence, enzymes are worthy targets for better under- standing pathogenesis and identifying new options for combatting fungal infections. Advances in genomics, proteomics, tran- scriptomics, and mass spectrometry have enabled the identification and characterization of new fungal enzymes. This review focuses on recent developments in the virulence-associated enzymes from Cryptococcus neoformans. The enzymatic suite of C. neoformans has evolved for environmental survival, but several of these enzymes play a dual role in colonizing the mammalian host. We also discuss new therapeutic and diagnostic strategies that could be based on the underlying enzymology. T he facultative intracellular fungal pathogen Cryptococcus neo- formans is the causative agent of cryptococcosis, a disease that primarily affects individuals with impaired immunity, such as those with advanced HIV infection (1, 2). C. neoformans is a ubiq- uitous environmental fungus associated with both pigeon guano and eucalyptus trees, and its environmental niche ranges from the tropical to the temperate (3). C. neoformans infection is acquired from the environment via inhalation, after which it forms a local infection in the lungs. This infection may be cleared, may be con- tained as a granuloma, or may disseminate from this initial site, leading to pneumonia and/or meningoencephalitis, the latter be- ing uniformly fatal if untreated. Despite the availability of antifun- gal therapy, more than 650,000 people die each year from C. neo- formans infection (1, 2, 4). The principal virulence factors of C. neoformans are a polysaccharide capsule, melanin production (5, 6), the ability to grow at body temperature (7), and the secretion of extracellular enzymes (7). These virulence factors confer a selec- tive advantage to C. neoformans for both residing in the environ- ment and in a mammalian host. Tightly controlled regulation leads to expression of enzymes required for fungal survival and host damage once inside its mammalian host (8). Many enzymes contribute to the composite cryptococcal viru- lence phenotype. Dissection of the pathogenic role of these en- zymes will enhance our understanding of cryptococcal pathogenic mechanisms and facilitate directed inhibitor development and/or vaccine discovery. We have included a table summarizing basic information regarding global C. neoformans enzymology (Table 1) and a schematic displaying localization of most of the high- lighted enzymes discussed (Fig. 1). In this review, we discuss in detail the most important virulence-associated enzymes (Table 2), as well as additional target enzymes with potential for rational antifungal drug design (Table 3). We examine this information in the context of infection and analyze candidate target enzymes for drug inhibition and vaccine discovery. POLYSACCHARIDE CAPSULE C. neoformans is the only fungal pathogen with a polysaccharide capsule, an outermost polysaccharide structure located just out- side the cell wall. The two major polysaccharide capsule constitu- ents are glucuronoxylomannan (GXM) and glucuroxyloman- nogalactan (GXMGal) (9–11). GXM is the major component of C. neoformans, a compound of -1,3-linked mannose residues with xylosyl and glucuronyl side groups (12), whereas GXMGal is made of -1,6-linked galactose residues with xylose, mannose, and glucuronic acid (13). The capsule also contains nonpolysaccha- ride components, such as mannoprotein (MP) (10, 14, 15), al- though these MP components may represent transient compo- nents destined for cellular export. The role of capsule in environmental growth is unknown, al- though speculations have been made that the capsule protects the fungus from desiccation or acts as a food source (16). During mammalian infection, the capsule participates in resisting phago- cytosis and modulating the immune response (17–21). Not only protective against phagocytosis in both mammalian and lepi- dopteran hosts (22, 23), the capsule also protects the fungus after ingestion by serving as a free radical sink that can shield the cell from oxidative bursts (24). Hence, while the capsule is not part of the enzymatic microbial arsenal, the machinery responsible for capsule synthesis and assembly does directly contribute to crypto- coccal virulence. The primary structures of GXM and GXMGal subunits have been defined, but the mechanisms of subunit as- sembly into 10 6 -Da branched structures have not (25, 26). The degree of branching and conformation of polysaccharides imply an elaborate assembly and regulatory enzymatic machinery (27). The subunits of GXM and GXMGal are large glycans that re- quire several glycosyltransferases for synthesis. Both xylosyltrans- ferase and glucuronyltransferase activities are involved in capsular polysaccharide biosynthesis (28–31). A xylosyltransferase, Cxt1, was the first glycosyltransferase identified with a defined role in capsule synthesis (31). It is a large transmembrane protein with -1,2-xylosyltransferase activity (31), and deletion of the corre- sponding gene (CXT1) decreased capsular -1,2-xylose linkages and fungal growth in the lung in a mouse model of infection (30). Several acapsular mutants were obtained through identifica- Accepted manuscript posted online 9 October 2015 Citation Almeida F, Wolf JM, Casadevall A. 2015. Virulence-associated enzymes of Cryptococcus neoformans. Eukaryot Cell 14:1173–1185. doi:10.1128/EC.00103-15. Address correspondence to Arturo Casadevall, [email protected]. Copyright © 2015, American Society for Microbiology. All Rights Reserved. MINIREVIEW December 2015 Volume 14 Number 12 ec.asm.org 1173 Eukaryotic Cell on January 3, 2020 by guest http://ec.asm.org/ Downloaded from

Transcript of Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in...

Page 1: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

Virulence-Associated Enzymes of Cryptococcus neoformans

Fausto Almeida,a,b Julie M. Wolf,a Arturo Casadevalla,c

Department of Microbiology and Immunology, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, USAa; Department of Cellular and MolecularBiology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Sao Paulo, Brazilb; Department of Molecular Microbiology and Immunology, Johns HopkinsBloomberg School of Public Health, Baltimore, Maryland, USAc

Enzymes play key roles in fungal pathogenesis. Manipulation of enzyme expression or activity can significantly alter the infec-tion process, and enzyme expression profiles can be a hallmark of disease. Hence, enzymes are worthy targets for better under-standing pathogenesis and identifying new options for combatting fungal infections. Advances in genomics, proteomics, tran-scriptomics, and mass spectrometry have enabled the identification and characterization of new fungal enzymes. This reviewfocuses on recent developments in the virulence-associated enzymes from Cryptococcus neoformans. The enzymatic suite of C.neoformans has evolved for environmental survival, but several of these enzymes play a dual role in colonizing the mammalianhost. We also discuss new therapeutic and diagnostic strategies that could be based on the underlying enzymology.

The facultative intracellular fungal pathogen Cryptococcus neo-formans is the causative agent of cryptococcosis, a disease that

primarily affects individuals with impaired immunity, such asthose with advanced HIV infection (1, 2). C. neoformans is a ubiq-uitous environmental fungus associated with both pigeon guanoand eucalyptus trees, and its environmental niche ranges from thetropical to the temperate (3). C. neoformans infection is acquiredfrom the environment via inhalation, after which it forms a localinfection in the lungs. This infection may be cleared, may be con-tained as a granuloma, or may disseminate from this initial site,leading to pneumonia and/or meningoencephalitis, the latter be-ing uniformly fatal if untreated. Despite the availability of antifun-gal therapy, more than 650,000 people die each year from C. neo-formans infection (1, 2, 4). The principal virulence factors of C.neoformans are a polysaccharide capsule, melanin production (5,6), the ability to grow at body temperature (7), and the secretion ofextracellular enzymes (7). These virulence factors confer a selec-tive advantage to C. neoformans for both residing in the environ-ment and in a mammalian host. Tightly controlled regulationleads to expression of enzymes required for fungal survival andhost damage once inside its mammalian host (8).

Many enzymes contribute to the composite cryptococcal viru-lence phenotype. Dissection of the pathogenic role of these en-zymes will enhance our understanding of cryptococcal pathogenicmechanisms and facilitate directed inhibitor development and/orvaccine discovery. We have included a table summarizing basicinformation regarding global C. neoformans enzymology (Table1) and a schematic displaying localization of most of the high-lighted enzymes discussed (Fig. 1). In this review, we discuss indetail the most important virulence-associated enzymes (Table 2),as well as additional target enzymes with potential for rationalantifungal drug design (Table 3). We examine this information inthe context of infection and analyze candidate target enzymes fordrug inhibition and vaccine discovery.

POLYSACCHARIDE CAPSULE

C. neoformans is the only fungal pathogen with a polysaccharidecapsule, an outermost polysaccharide structure located just out-side the cell wall. The two major polysaccharide capsule constitu-ents are glucuronoxylomannan (GXM) and glucuroxyloman-nogalactan (GXMGal) (9–11). GXM is the major component of C.

neoformans, a compound of �-1,3-linked mannose residues withxylosyl and glucuronyl side groups (12), whereas GXMGal is madeof �-1,6-linked galactose residues with xylose, mannose, andglucuronic acid (13). The capsule also contains nonpolysaccha-ride components, such as mannoprotein (MP) (10, 14, 15), al-though these MP components may represent transient compo-nents destined for cellular export.

The role of capsule in environmental growth is unknown, al-though speculations have been made that the capsule protects thefungus from desiccation or acts as a food source (16). Duringmammalian infection, the capsule participates in resisting phago-cytosis and modulating the immune response (17–21). Not onlyprotective against phagocytosis in both mammalian and lepi-dopteran hosts (22, 23), the capsule also protects the fungus afteringestion by serving as a free radical sink that can shield the cellfrom oxidative bursts (24). Hence, while the capsule is not part ofthe enzymatic microbial arsenal, the machinery responsible forcapsule synthesis and assembly does directly contribute to crypto-coccal virulence. The primary structures of GXM and GXMGalsubunits have been defined, but the mechanisms of subunit as-sembly into �106-Da branched structures have not (25, 26). Thedegree of branching and conformation of polysaccharides implyan elaborate assembly and regulatory enzymatic machinery (27).

The subunits of GXM and GXMGal are large glycans that re-quire several glycosyltransferases for synthesis. Both xylosyltrans-ferase and glucuronyltransferase activities are involved in capsularpolysaccharide biosynthesis (28–31). A xylosyltransferase, Cxt1,was the first glycosyltransferase identified with a defined role incapsule synthesis (31). It is a large transmembrane protein with�-1,2-xylosyltransferase activity (31), and deletion of the corre-sponding gene (CXT1) decreased capsular �-1,2-xylose linkagesand fungal growth in the lung in a mouse model of infection (30).

Several acapsular mutants were obtained through identifica-

Accepted manuscript posted online 9 October 2015

Citation Almeida F, Wolf JM, Casadevall A. 2015. Virulence-associated enzymes ofCryptococcus neoformans. Eukaryot Cell 14:1173–1185. doi:10.1128/EC.00103-15.

Address correspondence to Arturo Casadevall, [email protected].

Copyright © 2015, American Society for Microbiology. All Rights Reserved.

MINIREVIEW

December 2015 Volume 14 Number 12 ec.asm.org 1173Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 2: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

TABLE 1 Described enzymes in Cryptococcus neoformans

Enzyme Function(s)a EC no. Reference(s)

Localized on capsule and/or cell wall1,3-�-Glucan synthase Involved in �-glucan synthesis 2.4.1.34 135Acid phosphatase Involved in fungal cell adhesion to host tissues, localized in lysosomes,

and related to virulence (Table 2)3.1.3.2 106, 136, 137

Cas1 glycosyltransferase Participates in O-acetylation 2.4.1.X 138Chitin deacetylase Involved in chitin metabolism 3.5.1.41 139Chitin synthase Involved in chitin synthesis 2.4.1.16 140Chitinase Involved in chitin degradation 3.2.1.14 141Creatinine deaminase Involved in arginine and proline metabolism 3.5.4.21 142Esterase lipase Catalyzes hydrolysis of fatty acids 3.1.1.3 136GDP-mannose pyrophosphorylase Involved in GDP-mannose synthesis 2.7.7.13 143Glucan 1,3-�-glucosidase Involved in glucan synthesis 3.2.1.58 16Glucan 1,4-�-glucosidase Involved in glucan synthesis 3.2.1.3 16Gmt1 GDP-mannose Transport of GDP-mannose 2.7.7.22 144Lactonohydrolase Deficient strains show larger capsule size and facilitated immune

evasion3.1.1.15 37

N-Acetylgalactosaminoglycan deacetylase Involved in polysaccharide metabolism 3.1.1.58 145Phosphoaminase Involved in amino acid synthesis 136Phosphomannomutase Involved in GDP-mannose synthesis 5.4.2.8 143Phosphomannose isomerase Involved in GDP-mannose synthesis 5.3.1.8 143Uph1 ATPase Required for vesicle acidification 146Uxs1 decarboxylase Converts UDP-glucuronic acid to UDP-xylose 147�-1,3-Glucanase Involved in glucan synthesis 3.2.1.59 16�-Amylase Hydrolyzes alpha bonds of several polysaccharides and involved in cell

wall building3.2.1.1 148

�-Glucosidase Breaks down disaccharides to glucose and starch and involved in cellwall building

3.2.1.20 136

�-Mannosidase Involved in cell building through mannose metabolism 3.2.1.24 136�-Mannosyltransferase Involved in polysaccharide metabolism 2.4.1.132 38, 149�-Endoglucanase Involved in cell wall formation 3.2.1.4 148�-Glucosidase Involved in cell wall formation 3.2.1.21 136�-Glucuronidase Involved in cell wall formation, catalyzing breakdown of complex

carbohydrates3.2.1.31 136

Secreted/releasedAcyltransferase Involved in food acquisition 3.1.1.3 92Alkaline phosphatase Involved in regulation of signaling cascades and several protein

structure and localized in endoplasmic reticulum3.1.3.1 150

Aspartyl protease Involved in food acquisition 3.4.23.X 111Cellulase Involved in polysaccharide degradation 3.2.1.4 151DNase DNA degradation and related to virulence (Table 2) 3.1.21.1 79Metalloprotease Catalyzes mechanism that involves a metal and related to virulence

(Table 2)3.4.24.77 113, 152

Phospholipase B Similar to phospholipase C function, degrades cell membranecomponents, supports fungal attachment to host cells, localized oncell wall, and related to virulence (Table 2)

3.1.1.5 91, 92

Phospholipase C Degrades cell membrane components, supports fungal attachment tohost cells, and related to virulence (Table 2)

3.1.4.11 93

Protease Performs proteolysis interfering with host defense response 3.4.21.53 107, 108S2P endopeptidase Performs proteolysis 3.4.24.85 153Serine peptidase Performs proteolysis, coordinating several physiological functions 3.4.21.X 152Superoxide dismutase Catalyzes dismutation of toxic superoxide, converting superoxide to

hydrogen peroxide and oxygen and related to virulence (Table 2)1.15.1.1 83-85

Localized intracellularly2-Methylcitrate synthase Converts acyl groups into alkyl groups on transfer 2.3.3.5 1543-�-Hydroxysteroid 3-dehydrogenase Oxidizes a substrate by reduction reaction that transfers 1 or more

hydrides to electron acceptor1.1.1.270 155

6-Phosphogluconate dehydrogenase Involved in production of ribulose 1.1.1.44 156, 157Acetate kinase Catalyzes formation of acetyl-CoA 2.7.2.1 158Aconitase Catalyzes isomerization of citrate to isocitrate and involved in

response to nitrosative stress4.2.1.3 159

(Continued on following page)

Minireview

1174 ec.asm.org December 2015 Volume 14 Number 12Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 3: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

TABLE 1 (Continued)

Enzyme Function(s)a EC no. Reference(s)

Adenylyl cyclase Cac1 Converts ATP to cAMP 4.6.1.1 160Alternative oxidase Part of electron transport chain in mitochondria 1.10.3.11 161Aminopeptidase Catalyzes cleavage of amino acids from amino terminus of protein 3.4.11.21 137C-9-methyltransferase Involved in glycosphingolipid pathway 2.1.1.129 127Can2 carbonic anhydrase Responds directly to intracellular carbon oxide 4.2.1.1 162, 163Casein kinase 1 Dephosphorylation of Hog1 under stress conditions 2.7.11.1 164Catalase Protects cells from oxidative damage by reactive oxygen species 1.11.1.6 137, 150Cytochrome c peroxidase Takes reduced equivalents from cytochrome c and reduces hydrogen

peroxide to water1.11.1.5 165

Deacetylase Removes acetyl groups from lysine in proteins and is localized in cellwall

3.5.1.108 166

Dolichyl-diphosphooligosaccharide-proteinglycotransferase

Participates in N-glycan biosynthesis 2.4.99.18 167

Ferrochelatase Catalyzes final step in heme biosynthesis from highly photoreactiveporphyrins

4.99.1.1 168

Flippase Participates in phospholipid translocation between membrane sidesand localized in cell wall

3.6.3.1 169, 170

Glucose-6-phosphate dehydrogenase Is in pentose phosphate pathway, maintaining the level of coenzymeNADPH

1.1.1.49 171

Glucose-phosphate isomerase Catalyzes conversion of glucose-6-phosphate into fructose6-phosphate

5.3.1.9 172

Glucosylceramide synthase Involved in glucosylceramide synthesis, localized in cell wall, andrelated to virulence (Table 2)

2.4.1.80 127, 128

Glucuronyltransferase Involved in biosynthetic pathway of O-acetylated mannan 2.4.1.17 28Glutathione peroxidase Protects cells from oxidative damage 1.11.1.9 173Glyoxal oxidase Copper metalloenzyme that catalyzes oxidation of aldehydes to

corresponding carboxylic acids coupled to reduction of dioxygen toH2O2

1.2.1.23 148

Homoisocitrate dehydrogenase Participates in lysine biosynthesis 1.1.1.87 115Homoserine kinase Participates in glycine, serine, and threonine metabolism 2.7.1.39 174Homoserine O-acetyltransferase Participates in methionine and sulfur metabolism 2.3.1.31 175Hyaluronic synthase Involved in production of glycosaminoglycan at cell surface 2.4.1.212 176Imidazole glycerol-phosphate dehydratase Participates in histidine biosynthesis 4.2.1.19 177IMP dehydrogenase Participates in GTP biosynthesis 1.1.1.205 178Inositol phosphotransferase 1 Involved in glycosphingolipid pathway 2.7.1.X 127Inositol-phosphorylceramide synthase Involved in glycosphingolipid pathway 2.7.1.X 179Ire1 kinase Involved in cellular response to unfolded proteins 2.7.11.1 180Isocitrate lyase Catalyzes cleavage of isocitrate to succinate and glyoxylate 4.1.3.1 181Laccase Polyphenol oxidase and copper-containing oxidase enzyme, localized

in cell wall, and related to virulence (Table 2)1.10.3.2 45, 46, 50

Malate dehydrogenase Catalyzes oxidation of malate to oxaloacetate 1.1.1.37 182Mannitol-1-phosphate 5-dehydrogenase Participates in fructose and mannose metabolism 1.1.1.17 183, 184Mannose-1-phosphate guanylyltransferase(GDP)

Participates in fructose and mannose metabolism 2.7.7.22 144

Mannosyl phosphorylinositol ceramidesynthase

Involved in glycosphingolipid pathway 2.4.X.X 127

Mannosyltransferase Participates in O-mannosylation of proteins and involved in cell wallintegrity and morphogenesis

2.4.1.109 185

Myristoyl-CoA: protein N-myristoyltransferase Catalyzes transfer of myristate from CoA to proteins 2.3.1.97 116Pde1 phosphodiesterase Modulates cAMP 3.1.4.1 186Phosphoglucomutase Participates in interconversion of glucose 1-phosphate and glucose

6-phosphate5.4.2.2 172

Protein farnesyltransferase Participates in formation of farnesyl protein and diphosphate 2.5.1.58 187Rho1 GTPase Involved in MAPK cascade 3.6.5.2 188RNase III Binds and cleaves double-stranded RNA 3.1.26.3 189Saccharopine dehydrogenase Participates in lysine metabolism 1.5.1.10 190Sphingolipid methyltransferase 1 Participates in methylation of glucosylceramide 2.1.1.1 191Sterol 14�-demethylase Involved in sterol metabolism 1.14.13.7 192Sterol 24-C-methyltransferase Involved in sterol metabolism 1.15.1.1 193Thiol peroxidase Reduces peroxides and inhibits hydrogen peroxide response 1.11.1.7 194

(Continued on following page)

Minireview

December 2015 Volume 14 Number 12 ec.asm.org 1175Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 4: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

tion of rough colonies. This type of screen identified four genesrequired for capsule formation: CAP10, CAP59, CAP60, andCAP64. Although these genes are not essential, their mutationdoes confer defects in growth and in mouse models of infection(17, 32–35). Cells from these mutant strains lacked or produced

extremely reduced capsule, but these mutations did not corre-late with enzymatic deficiency in UDP-glucose dehydrogenase,UDP-glucuronate decarboxylase, UDP-glucuronyl:acceptortransferase, UDP-xylosyl:acceptor transferase, or lipid-linkedoligosaccharide biosynthetic pathways. CAP10 is a putative xy-

TABLE 1 (Continued)

Enzyme Function(s)a EC no. Reference(s)

Thioredoxin reductase Catalyzes reduction of thioredoxin 1.8.1.9 195Threonine synthase Participates in glycine, serine, and threonine metabolism 4.2.3.1 174Thymidylate synthase Catalyzes conversion of dUMP to deoxythymidine monophosphate 2.1.1.45 196Transaldolase Involved in pentose phosphate pathway 2.2.1.2 159Trehalose-6-phosphate phosphatase Participates in starch and sucrose metabolism 3.1.3.12 197Trehalose-6-phosphate synthase Participates in starch and sucrose metabolism 2.4.1.15 197UDP-galactopyranose mutase Catalyzes conversion of UDP-D-galactopyranose in

UDP-D-galacto-1,4-furanose5.4.99.9 198

UDP-glucose dehydrogenase Participates in conversion of UDP-glucose to UDP-glucuronate, andformation of glycosaminoglycans

1.1.1.22 199

UDP-glucuronate decarboxylase Participates in nucleotide sugar metabolism 4.1.1.35 147Urease Catalyzes hydrolysis of urea into carbono dioxide and ammonia and

related to virulence (Table 2)3.5.1.5 74

Xylosylphosphotransferase Participates in O-glycosylation biosynthesis and related to virulence(Table 2)

2.7.8.32 28, 31, 200

�8 desaturase Involved in glycosphingolipid pathway 1.14.19.4 127a cAMP, cyclic AMP; MAPK, mitogen-activated protein kinase.

FIG 1 Enzymes are crucial for fungal pathogenesis and can alter the infection process. These enzymes are potential targets for new antifungal agents. (A) Somepathogenesis-related enzymes are retained to be active inside the cell body, while others are secreted. Some, like laccase, are both retained and secreted. (B) Ofthose released, some are secreted using traditional secretion systems, while others are included as cargo in extracellular vesicles.

Minireview

1176 ec.asm.org December 2015 Volume 14 Number 12Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 5: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

losyltransferase gene, and cap10� mutants show a pleiotropicphenotype, which includes enlarged cell size, smaller extracel-lular vesicles, and affected expression of some virulence factors(36). CAP10 therefore is required for both capsule formationand other aspects of fungal virulence.

Capsular lactonohydrolase also affects multiple capsule-re-lated phenotypes (37). A strain lacking lactonohydrolase (lhc1�)produced capsules with a larger size and altered branching, den-sity, and solvation compared to the parental strain. These capsularstructure alterations increased virulence in murine infection (37).Taken together, these results suggest that lactone may be involvedin cross-linking of the capsule.

�-1,3-Mannosyltransferase (encoded by CMT1) synthesizesthe mannose backbone of GXM and thus plays a crucial role incapsule synthesis. However, �-1,3-mannosyltransferase activity ismore involved in in serotype A capsule biosynthesis than in theserotype D C. neoformans (38, 39). Serotypes A and D representtwo of the four C. neoformans serotypes: C. neoformans var. neo-formans (serotypes A and D) and C. neoformans var. gattii (sero-types B and C), which can be distinguished according to theirgrowth differences on diagnostic media (40). The strain-specificcapsule synthesis differences, such as the role of CMT1, show theimportance of studying multiple strain backgrounds.

Much remains to be learned about the enzymatic machineryinvolved in capsule synthesis, including enzyme localization andkinetics. Detailed studies of capsule structure and the enzymaticmachinery involved are critical for a better understanding of thefunction of the capsule production and regulation.

MELANIN SYNTHESIS

Melanin formation protects C. neoformans from oxidative damageas well as from both heat and cold (41, 42). Melanin is synthesized

on 2,3- or 3,4-diphenol substrates by a phenoloxidase and accu-mulates in the C. neoformans cell wall (43, 44). The melanin-syn-thesizing enzyme has two classical laccase characteristics: a glyco-sylated copper-containing protein with the ability to oxidizediphenolic substrates and the ability to produce decarboxy dop-achrome (45, 46). C. neoformans melanin synthesis occurs only inthe presence of exogenous dihydroxyphenols, since no known C.neoformans endogenous substrate exists. Several diphenols canserve as the substrates for pigment synthesis by C. neoformanslaccase (47), such as the substrates consisting of para- and ortho-diphenols, monophenols, L-dopa, and esculin, indicating that theenzyme has broad specificity and the ability to generate pigmentsfrom different compounds (47–53). Iron increases laccase activ-ity, but hydrogen peroxide has no effect on enzymatic activity,despite the antioxidant properties of melanin (54).

The genes LAC1 and LAC2 encode two laccases, but a singledeletion in LAC1 is able to prevent melanin production (55–58).Lac1 localizes in the cell wall, while Lac2 is cytoplasmic, but Lac2can localize to the cell wall in the absence of Lac1 (55). lac1�mutants are easily identified as white colonies when cultivated oncatecholamine-containing media (59). The lac1� mutant showsdecreased virulence in survival studies with rabbit infection (59),corroborating the important role in the fungal virulence (5, 46). Inaddition to its cell wall localization, laccase is packaged into extra-cellular vesicles, a nontraditional mechanism of secretion, and cantherefore mediate damage away from the laccase-producing fun-gal cell (Fig. 1).

Melanin is considered a powerful antioxidant, since it mayprotect cryptococcal cells against oxygen- and nitrogen-derivedoxidants of the type made by host effector cells (5, 60–62). Inaddition to its capacity to absorb free radical fluxes, melanin canalso contribute to acquired resistance against to the antifungals

TABLE 2 Enzymes related to the virulence in Cryptococcus neoformans

Enzyme Comment(s) Reference(s)

Acid phosphatase Deficient strains show affected virulence in mouse and Galleria mellonella models of infection 106DNase Acts in degrading host DNA and supplies C. neoformans with nucleotides 79Glucosylceramide synthase Required for virulence in murine model of infection 127, 128Laccase Deficient strains show decreased virulence in survival studies with rabbit and mouse models of infection 59Mannosyltransferase Required for virulence in murine model of infection 185Metalloprotease Deficient strains unable to cross endothelium in in vitro model of human blood-brain barrier and is

required for invasion of central nervous system113

Phospholipase B Required in invasion of host tissue and dissemination in murine model 95Phospholipase C Shown to be important for several virulence phenotypes 101, 102Superoxide dismutase Attenuated growth of deficient strains within macrophages 89Urease Deficient strains less virulent than wild-type strain in mouse model of infection and is involved in

fungal escape from lung to cross blood-brain barrier76

Xylosylphosphotransferase Deficient strains manifest reduced growth in lung tissue in mouse model of infection 30

TABLE 3 Possible target enzymes for rational antifungal drug design

Enzyme(s) Comment(s) Reference(s)

14�-Demethylase A critical enzyme in sterol assembly 119Glucosylceramide synthase Glucosylceramide plays critical role in pathogenicity of C. neoformans 127, 128Laccase Melanization aids virulence 60, 63, 64, 65Myristoyltransferase Myristoylation inhibition is fatal for C. neoformans 116, 117Phosphoribosylaminoimidazole carboxylase Mutants that cannot synthesize adenine have reduced virulence 114Pyrophosphorylase and cytosine-specific permease Enzymes are basis of C. neoformans flucytosine resistance 201, 202Sterol synthesis enzymes Sterol synthesis enzyme mutants show resistance to fluconazole and amphotericin 122-124

Minireview

December 2015 Volume 14 Number 12 ec.asm.org 1177Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 6: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

amphotericin B and caspofungin, since nonmelanized cryptococ-cal cells are more susceptible than melanized cells to amphotericinB and caspofugin. Moreover, killing assays demonstrated that ad-dition of melanin particles to amphotericin B or caspofungin sig-nificantly reduces their toxicities against C. neoformans (63–65).Thus, melanin and laccase are considered promising targets fordrugs against C. neoformans infection.

EXTRACELLULAR ENZYMES

As nature’s “recyclers,” environmental fungi secrete a number ofdegradative enzymes to breakdown macromolecules and obtainnutrients in the environment (7, 66–69). C. neoformans is no ex-ception and releases a number of lipases, proteases, and DNases.However, during the infection process, the same degradative en-zymes contribute to virulence by destroying tissues, promotingfungal survival, and interfering with effective immune responses.

Urease is almost universally expressed by C. neoformans iso-lates. In the environment, C. neoformans is often isolated fromavian excreta (70, 71). To survive and grow on this medium, thefungus must metabolize creatinine, xanthines, and uric acid. Highurease activity may benefit the fungus under these conditions (72–74), as the enzyme catalyzes the hydrolysis of urea to ammoniaand carbamate. Urease is considered a major cryptococcal viru-lence factor (75). A urease knockout (URE1) strain of C. neofor-mans was significantly less virulent than the wild-type strain in amouse model of infection (76). Urease plays a role in fungal escapefrom the lung to cross the blood-brain barrier but is not requiredfor fungal growth once inside the brain (76). Urease productionvaries among clinical isolates; however, the vast majority (99.6%)demonstrate some level of urease activity (74, 77, 78). Neverthe-less, occasional urease-negative variants have been isolated in clin-ical isolates (77), suggesting that this enzyme can be dispensable,provided that there are compensatory virulence mechanisms.

Extracellular DNase is produced by C. neoformans in highquantities (79). This DNase may degrade host DNA secreted byneutrophils as part of the innate immune response (80) and addi-tionally may supply C. neoformans with nucleotides. A survey ofseveral yeast species, including C. neoformans, suggests a correla-tion between urease activity and extracellular DNase production(79). DNase activity is stronger in clinical strains than in environ-mental strains, further suggesting DNase may play a role as a vir-ulence factor (81).

Superoxide dismutases (SODs) convert superoxide to hydro-gen peroxide and oxygen (82). Two SODs have been described inC. neoformans (83–88). SOD contributes to virulence of C. neofor-mans by facilitating growth within macrophages (89), through amechanism that is likely to involve protection of the fungusagainst superoxide generated by host immune response (2). In thisregard, melanin and SOD may stimulate complementary defensesfor the C. neoformans cells’ protection against oxidative damage.SOD production is regulated by temperature, with increases inexpression at 37°C compared to 25°C. Thus, increased SOD pro-duction at body temperatures may protect the fungus against ox-idizing agents produced from host effector cells (90).

Phospholipases degrade cell membrane phospholipids in anenzyme-dependent mechanism. C. neoformans extracellular su-pernatants contain phospholipase B, phospholipase C, lysophos-pholipase, and acyltransferase (91–93), and phospholipase activ-ity supports fungal attachment to host cells (94). Phospholipase Bpromotes fungal invasion of host tissue (95) and hydrolyzes phos-

pholipids in lung surfactant and the plasma membrane (92, 96).Moreover, it contributes to fungal survival by maintaining cellwall integrity (97) and provides nutrients that can be used as solecarbon sources by C. neoformans during the infection (98, 99). Asdescribed above, it has also been localized to the cell wall (97), andits transport to the plasma membrane and cell wall is N-glycandependent (100). Phospholipase C is crucial for several virulencephenotypes (melanin production, growth at 37°C, phospholipaseB secretion, and antifungal drug resistance) and is also involved inhomeostasis regulation, cell separation following cytokinesis, andcell wall integrity (101, 102).

Phosphatases remove a phosphate group from their substratesand play important roles in regulating protein structure and sig-naling cascades (103, 104). A secreted acid phosphatase is involvedin fungal cell adhesion to host tissues, suggesting an importantrole in establishing infection (105). Acid phosphatase is encodedby the gene APH1 in C. neoformans. In both wax worm and mu-rine models of cryptococcosis, aph1� strain-infected animals sur-vived longer than those in the wild-type-infected model (106),demonstrating the importance of this enzyme during infection.

Proteases break down proteins and are considered importantvirulence factors, contributing to tissue invasion, colonization,and alteration of the host defense response. Protease activity in C.neoformans cultures has been reported by several investigators(107–111). Proteases play important roles in host cell penetrationand virulence of C. neoformans (112). Recently, a metalloproteasewas identified by proteomic analyses of the extracellular proteinsfrom C. neoformans and found to be required for invasion of thecentral nervous system in murine infection of C. neoformans(113). Moreover, the metalloprotease knockout (mpr1�) strainwas unable to cross the endothelium in an in vitro model of thehuman blood-brain barrier (113).

DRUG DESIGN AND RESISTANCE

Definition of enzymatic pathways can provide crucial targets forantimicrobial drug design. One way to identify targets is to iden-tify unique metabolic requirements for cryptococcal growthand/or virulence. An example of this is the C. neoformans phos-phoribosylaminoimidazole carboxylase gene (ADE2). Mutantswith mutations in this gene lack an enzyme required for adeninesynthesis and thus have reduced virulence compared to the wild-type strain (114). This observation suggests potential for rationaldrug design utilizing differences in adenine synthesis pathwaysbetween host and pathogen (as first suggested in reference 7).Several candidate enzymes in C. neoformans have been studiedregarding fungal amino acid synthesis (e.g., homocitrate synthase,homoisocitrate dehydrogenase, �-aminoadipate reductase, sac-charopine reductase, and saccharopine dehydrogenase) (115).However, comparisons between C. neoformans var. neoformansand C. neoformans var. gattii have shown that candidate targets donot necessarily translate across Cryptococcus species. Saccharopinereductase, an enzyme involved in lysine synthesis, was not de-tected in C. neoformans var. gattii but was detected in C. neofor-mans var. neoformans. This C. neoformans var. gattii strain wasable to grow even in the absence of lysine (115), indicating thatfurther research to identify enzymes essential across all Cryptococ-cus species is required.

Another essential process for C. neoformans is protein myris-toylation. C. neoformans myristoyltransferase catalyzes the trans-fer of myristate from coenzyme A (CoA) to the amino-terminal

Minireview

1178 ec.asm.org December 2015 Volume 14 Number 12Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 7: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

glycine residue of a subset of cellular proteins, and this enzyme isessential for C. neoformans viability (116, 117). N-Myristoyl pro-teins and myristoylation inhibition by the myristic acid analog4-oxatetradecanoic acid are crucial for this organism (118). Thus,therapies directed at myristoylation may also be a possible targetfor rational antifungal drug design.

In some cases, an antifungal target is well defined, but multipleenzymes involved in target synthesis provide several inhibitorystrategies. Sterols and their synthetic pathways are major antifun-gal targets in many fungi, but resistance leads to difficulties inpatient treatment. Fluconazole-resistant strains require a 10-fold-higher drug concentration to inhibit sterol 14�-demethylation(119), rendering the drug clinically unfeasible. The molecular ba-sis for differential enzyme function has been identified in severalclinical C. neoformans strains (120). One documented flucona-zole- and amphotericin-resistant C. neoformans patient isolateshowed reduced relative sterol content and a defect in �-8-isomer-ase, depleted ergosterol, and accumulated aberrant �-8-double-bonded ergosterol precursors (121, 122), suggesting the ability toform membrane pores due to aggregation and formation of am-photericin-ergosterol complexes. Another study evaluating flu-conazole- and amphotericin-resistant isolates observed reducedergosterol content in the isolates, as well as reduced sensitivity ofP450 14�-demethylase to inhibition by fluconazole, and a defectin sterol �8-�7 isomerase (123). Another C. neoformans strainwith defective sterol �8-�7 isomerase was discovered in an am-photericin B-resistant isolate from an AIDS patient (124). Thesemutations in sterol synthesis enzymes explain resistance evolutionand generate targets to fight it with. This information can also helpin rational drug design methodologies.

Identification of key virulence-related enzymes is yet anotherroute toward finding an effective drug target. Glycosphingolipidsare essential to regulate survival and/or replication of C. neofor-mans in the phagolysosome, as well as in the extracellular environ-ment of the host (125–127). Glucosylceramide plays critical role inpathogenicity of C. neoformans, since glucosylceramide synthase(Gcs1) is required for virulence in the murine model of infection(128). gcs1� mutants corroborate the crucial role of the glycosph-ingolipid synthesis in regulation of this considerable aspect of C.neoformans virulence (127). Thus, the glycosphingolipid pathwaymay also be a reasonable target for antifungal therapies.

Laccase has been considered a drug target in C. neoformansbecause melanization is critical to virulence. Inhibition of fungalmelanization in murine infection using the herbicide glyphosateprolonged average mouse survival. Glyphosate is an inhibitor ofboth the shikimate acid pathway and L-dopa polymerization(129). Thus, therapies directed at melanization may also be a po-tential target for antifungal drug design.

Occasionally, a drug proven to work on one microbial patho-gen will also be effective against another. This appears to be thecase with several viral medications. Drugs such as indinavir andoseltamivir inhibit human immunodeficiency virus (HIV) pro-tease or influenza virus neuraminidase, respectively, and demon-strate the impact an enzymatic inhibitor can have in the clinic(130, 131). The use of protease inhibitors has shown positive ef-fects on C. neoformans and Candida albicans infections, wheredrug treatment was associated with inhibition of fungal growthand proliferation in vitro (132, 133). These are likely inhibiting thefungal proteases, both cell associated and as part of the fungalsecretome.

CONCLUSION

Recent advances in genomics, proteomics, transcriptomics, andmass spectrometry have facilitated the identification and charac-terization of new fungal enzymes, including those specific to bothfungi and C. neoformans. These enzymes are required for manyimportant biological processes, including growth and infection.The importance of the secretome in cryptococcal pathogenesis isapparent from the fact that strain differences in secreted enzymescorrelate with their virulence (134). Nonetheless, important ques-tions remain. Future research on cryptococcal enzymology willnot only identify new enzymes and their roles during infection butalso pinpoint enzymatic targets for the development of antifungalagents.

ADDENDUM IN PROOF

There are, of course, many enzymes involved in signaling cas-cades, most of which were not discussed in this review. One suchenzyme is vital to stress response in C. neoformans and otherpathogenic fungi and thus merits a well-deserved mention: thecalcium-dependent phosphatase calcineurin (W. J. Steinbach, J. L.Reedy, R. A. Cramer, Jr., J. R. Perfect, J. Heitman, Nat Rev Micro-biol 5:418 – 430, 2008). This enzyme is required for growth in amammalian host and therefore is necessary to cause disease (A.Odom, S. Muir, E. Lim, D. L. Toffaletti, J. Perfect, J. Heitman,EMBO J 16:2576 –2589, 1997). Studies utilizing calcineurin inhib-itors for invasive disease in animal models have shown promisingresults, and this work is now moving into translational stages(D. P. Kontoyiannis, R. E. Lewis, B. D. Alexander, O. Lortholary,F. Dromer, K. L. Gupta, G. T. John, R. del Busto, G. B. Klintmalm,J. Somani, G. M. Lyon, K. Pursell, V. Stosor, P. Munoz, A. P.Limaye, A. C. Kalil, T. L. Pruett, J. Garcia-Diaz, A. Humar, S.Houston, A. A. House, D. Wray, S. Orloff, L. A. Dowdy, R. A.Fisher, J. Heitman, N. D. Albert, M. M. Wagener, N. Singh, Anti-microb Agents Chemother 52:735–738, 2008, http://dx.doi.org/10.1128/AAC.00990-07). Other enzymes involved in stress re-sponses may similarly be identified and targeted in the future.

REFERENCES1. Park BJ, Wannemuehler KA, Marston BJ, Govender N, Pappas PG,

Chiller TA. 2009. Estimation of the current global burden of cryptococ-cal meningitis among persons living with HIV/AIDS. AIDS 23:525–530.http://dx.doi.org/10.1097/QAD.0b013e328322ffac.

2. Heitman J, Kozel TR, Kwon-Chung J, Perfect JR, Casadevall A. 2011.Cryptococcus: from human pathogen to model yeast. ASM Press, Wash-ington, DC.

3. Nielsen K, De Obaldia AL, Heitman J. 2007. Cryptococcus neoformansmates on pigeon guano: implications for the realized ecological nicheand globalization. Eukaryot Cell 6:949 –959. http://dx.doi.org/10.1128/EC.00097-07.

4. Mitchell TG, Perfect JR. 1995. Cryptococcosis in the era of AIDS—100years after the discovery of Cryptococcus neoformans. Clin Microbiol Rev8:515–548.

5. Williamson PR. 1997. Laccase and melanin in the pathogenesis of Cryp-tococcus neoformans. Front Biosci 2:e99 – e107.

6. Casadevall A, Rosas AL, Nosanchuk JD. 2000. Melanin and virulence inCryptococcus neoformans. Curr Opin Microbiol 3:354 –358. http://dx.doi.org/10.1016/S1369-5274(00)00103-X.

7. Casadevall A, Perfect JR. 1998. Cryptococcus neoformans. ASM Press,Washington, DC.

8. Kronstad J, Saikia S, Nielson ED, Kretschmer M, Jung W, Hu G,Geddes JM, Griffiths EJ, Choi J, Cadieux B, Caza M, Attarian R. 2012.Adaptation of Cryptococcus neoformans to mammalian hosts: integratedregulation of metabolism and virulence. Eukaryot Cell 11:109 –118. http://dx.doi.org/10.1128/EC.05273-11.

Minireview

December 2015 Volume 14 Number 12 ec.asm.org 1179Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 8: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

9. Cherniak R, Reiss E, Turner SH. 1982. A galactoxylomannan antigen ofCryptococcus neoformans serotype A. Carbohydr Res 103:239 –250. http://dx.doi.org/10.1016/S0008-6215(00)80686-2.

10. Cherniak R, Sundstrom JB. 1994. Polysaccharide antigens of the capsuleof Cryptococcus neoformans. Infect Immun 62:1507–1512.

11. Bose I, Reese AJ, Ory JJ, Janbon G, Doering TL. 2003. A yeast undercover: the capsule of Cryptococcus neoformans. Eukaryot Cell 2:655– 663.http://dx.doi.org/10.1128/EC.2.4.655-663.2003.

12. Kozel TR, Levitz SM, Dromer F, Gates MA, Thorkildson P, Janbon G.2003. Antigenic and biological characteristics of mutant strains of Cryp-tococcus neoformans lacking capsular O acetylation or xylosyl side chains.Infect Immun 71:2868 –2875. http://dx.doi.org/10.1128/IAI.71.5.2868-2875.2003.

13. Heiss C, Klutts JS, Wang Z, Doering TL, Azadi P. 2009. The structureof Cryptococcus neoformans galactoxylomannan contains beta-D-glucuronic acid. Carbohydr Res 344:915–920. http://dx.doi.org/10.1016/j.carres.2009.03.003.

14. Jesus MD, Nicola AM, Chow SK, Lee IR, Nong S, Specht CA, LevitzSM, Casadevall A. 2010. Glucuronoxylomannan, galactoxylomannan,and mannoprotein occupy spatially separate and discrete regions in thecapsule of Cryptococcus neoformans. Virulence 1:500 –508. http://dx.doi.org/10.4161/viru.1.6.13451.

15. Rodrigues ML, Nimrichter L. 2012. In good company: association be-tween fungal glycans generates molecular complexes with unique func-tions. Frontiers Microbiol 3:249.

16. O’Meara TR, Alspaugh JA. 2012. The Cryptococcus neoformans capsule:a sword and a shield. Clin Microbiol Rev 25:387– 408. http://dx.doi.org/10.1128/CMR.00001-12.

17. Chang YC, Kwon-Chung KJ. 1994. Complementation of a capsule-deficient mutation of Cryptococcus neoformans restores its virulence. MolCell Biol 14:4912– 4919. http://dx.doi.org/10.1128/MCB.14.7.4912.

18. Rodrigues ML, Alviano CS, Travassos LR. 1999. Pathogenicity of Cryp-tococcus neoformans: virulence factors and immunological mechanisms.Microbes Infect 1:293–301. http://dx.doi.org/10.1016/S1286-4579(99)80025-2.

19. Kozel TR, Pfrommer GS, Guerlain AS, Highison BA, Highison GJ.1988. Role of the capsule in phagocytosis of Cryptococcus neoformans.Rev Infect Dis 10(Suppl 2):S436 –S439. http://dx.doi.org/10.1093/cid/10.Supplement_2.S436.

20. Pericolini E, Cenci E, Monari C, De Jesus M, Bistoni F, Casadevall A,Vecchiarelli A. 2006. Cryptococcus neoformans capsular polysaccharidecomponent galactoxylomannan induces apoptosis of human T-cellsthrough activation of caspase-8. Cell Microbiol 8:267–275. http://dx.doi.org/10.1111/j.1462-5822.2005.00619.x.

21. Vecchiarelli A, Monari C. 2012. Capsular material of Cryptococcus neo-formans: virulence and much more. Mycopathologia 173:375–386. http://dx.doi.org/10.1007/s11046-011-9513-8.

22. Trevijano-Contador N, Herrero-Fernandez I, Garcia-Barbazan I,Scorzoni L, Rueda C, Rossi SA, Garcia-Rodas R, Zaragoza O. 2015.Cryptococcus neoformans induces antimicrobial responses and behaves asa facultative intracellular pathogen in the non mammalian model Galle-ria mellonella. Virulence 6:66 –74. http://dx.doi.org/10.4161/21505594.2014.986412.

23. Alvarez M, Burn T, Luo Y, Pirofski LA, Casadevall A. 2009. Theoutcome of Cryptococcus neoformans intracellular pathogenesis in hu-man monocytes. BMC Microbiol 9:51. http://dx.doi.org/10.1186/1471-2180-9-51.

24. Zaragoza O, Rodrigues ML, De Jesus M, Frases S, Dadachova E,Casadevall A. 2009. The capsule of the fungal pathogen Cryptococcusneoformans. Adv Appl Microbiol 68:133–216. http://dx.doi.org/10.1016/S0065-2164(09)01204-0.

25. Frases S, Pontes B, Nimrichter L, Viana NB, Rodrigues ML, CasadevallA. 2009. Capsule of Cryptococcus neoformans grows by enlargement ofpolysaccharide molecules. Proc Natl Acad Sci U S A 106:1228 –1233.http://dx.doi.org/10.1073/pnas.0808995106.

26. McFadden DC, De Jesus M, Casadevall A. 2006. The physical proper-ties of the capsular polysaccharides from Cryptococcus neoformans sug-gest features for capsule construction. J Biol Chem 281:1868 –1875. http://dx.doi.org/10.1074/jbc.M509465200.

27. Cordero RJ, Frases S, Guimaraes AJ, Rivera J, Casadevall A. 2011.Evidence for branching in cryptococcal capsular polysaccharides andconsequences on its biological activity. Mol Microbiol 79:1101–1117.http://dx.doi.org/10.1111/j.1365-2958.2010.07511.x.

28. White CW, Cherniak R, Jacobson ES. 1990. Side group addition byxylosyltransferase and glucuronyltransferase in biosynthesis of capsularpolysaccharide in Cryptococcus neoformans. J Med Vet Mycol 28:289 –301. http://dx.doi.org/10.1080/02681219080000381.

29. Castle SA, Owuor EA, Thompson SH, Garnsey MR, Klutts JS,Doering TL, Levery SB. 2008. �1,2-Xylosyltransferase Cxt1p is solelyresponsible for xylose incorporation into Cryptococcus neoformans gly-cosphingolipids. Eukaryot Cell 7:1611–1615. http://dx.doi.org/10.1128/EC.00458-07.

30. Klutts JS, Doering TL. 2008. Cryptococcal xylosyltransferase 1 (Cxt1p)from Cryptococcus neoformans plays a direct role in the synthesis of cap-sule polysaccharides. J Biol Chem 283:14327–14334. http://dx.doi.org/10.1074/jbc.M708927200.

31. Klutts JS, Levery SB, Doering TL. 2007. A beta-1,2-xylosyltransferasefrom Cryptococcus neoformans defines a new family of glycosyltrans-ferases. J Biol Chem 282:17890 –17899. http://dx.doi.org/10.1074/jbc.M701941200.

32. Chang YC, Kwon-Chung KJ. 1999. Isolation, characterization, and lo-calization of a capsule-associated gene, CAP10, of Cryptococcus neofor-mans. J Bacteriol 181:5636 –5643.

33. Chang YC, Penoyer LA, KwonChung KJ. 1996. The second capsulegene of Cryptococcus neoformans, CAP64, is essential for virulence. InfectImmun 64:1977–1983.

34. Chang YC, Kwon-Chung KJ. 1998. Isolation of the third capsule-associated gene, CAP60, required for virulence in Cryptococcus neofor-mans. Infect Immun 66:2230 –2236.

35. Jacobson ES, Tingler MJ. 1994. Strains of Cryptococcus neoformans withdefined capsular phenotypes. J Med Vet Mycol 32:401– 404. http://dx.doi.org/10.1080/02681219480000531.

36. Tefsen B, Grijpstra J, Ordonez S, Lammers M, van Die I, de Cock H.2014. Deletion of the CAP10 gene of Cryptococcus neoformans results in apleiotropic phenotype with changes in expression of virulence factors.Res Microbiol 165:399 – 410. http://dx.doi.org/10.1016/j.resmic.2014.04.001.

37. Park YD, Shin S, Panepinto J, Ramos J, Qiu J, Frases S, AlbuquerqueP, Cordero RJ, Zhang N, Himmelreich U, Beenhouwer D, Bennett JE,Casadevall A, Williamson PR. 2014. A role for LHC1 in higher orderstructure and complement binding of the Cryptococcus neoformans cap-sule. PLoS Pathog 10:e1004037. http://dx.doi.org/10.1371/journal.ppat.1004037.

38. Doering TL. 1999. A unique alpha-1,3 mannosyltransferase of thepathogenic fungus Cryptococcus neoformans. J Bacteriol 181:5482–5488.

39. Sommer U, Liu H, Doering TL. 2003. An alpha-1,3-mannosyltransferase ofCryptococcus neoformans. J Biol Chem 278:47724 – 47730. http://dx.doi.org/10.1074/jbc.M307223200.

40. Bennett JE, Kwonchung KJ, Howard DH. 1977. Epidemiologic differ-ences among serotypes of Cryptococcus neoformans. Am J Epidemiol 105:582–586.

41. Rosas AL, Casadevall A. 1997. Melanization affects susceptibility ofCryptococcus neoformans to heat and cold. FEMS Microbiol Lett 153:265–272. http://dx.doi.org/10.1016/S0378-1097(97)00239-5.

42. Khajo A, Bryan RA, Friedman M, Burger RM, Levitsky Y, CasadevallA, Magliozzo RS, Dadachova E. 2011. Protection of melanized Crypto-coccus neoformans from lethal dose gamma irradiation involves changesin melanin’s chemical structure and paramagnetism. PLoS One6:e25092. http://dx.doi.org/10.1371/journal.pone.0025092.

43. Shaw CE, Kapica L. 1972. Production of diagnostic pigment by phe-noloxidase activity of Cryptococcus neoformans. Appl Microbiol 24:824 –830.

44. Wang Y, Aisen P, Casadevall A. 1996. Melanin, melanin “ghosts,” andmelanin composition in Cryptococcus neoformans. Infect Immun 64:2420 –2424.

45. Williamson PR. 1994. Biochemical and molecular characterization ofthe diphenol oxidase of Cryptococcus neoformans—identification as a lac-case. J Bacteriol 176:656 – 664.

46. Ikeda R, Shinoda T, Morita T, Jacobson ES. 1993. Characterization ofa phenol oxidase from Cryptococcus neoformans var. neoformans. Micro-biol Immunol 37:759 –764. http://dx.doi.org/10.1111/j.1348-0421.1993.tb01702.x.

47. Chaskes S, Tyndall RL. 1975. Pigment production by Cryptococcus neo-formans from para- and ortho-diphenols: effect of the nitrogen source. JClin Microbiol 1:509 –514.

Minireview

1180 ec.asm.org December 2015 Volume 14 Number 12Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 9: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

48. Edberg SC, Chaskes SJ, Alture-Werber E, Singer JM. 1980. Esculin-based medium for isolation and identification of Cryptococcus neofor-mans. J Clin Microbiol 12:332–335.

49. Kwon-Chung KJ, Tom WK, Costa JL. 1983. Utilization of indole com-pounds by Cryptococcus neoformans to produce a melanin-like pigment.J Clin Microbiol 18:1419 –1421.

50. Polacheck I, Hearing VJ, Kwon-Chung KJ. 1982. Biochemical studies ofphenoloxidase and utilization of catecholamines in Cryptococcus neofor-mans. J Bacteriol 150:1212–1220.

51. Polacheck I, Platt Y, Aronovitch J. 1990. Catecholamines and virulenceof Cryptococcus neoformans. Infect Immun 58:2919 –2922.

52. Strachan AA, Yu RJ, Blank F. 1971. Pigment production of Cryptococcusneoformans grown with extracts of Guizotia abyssinica. Appl Microbiol22:478 – 479.

53. Wang HS, Zeimis RT, Roberts GD. 1977. Evaluation of a caffeic acid-ferric citrate test for rapid identification of Cryptococcus neoformans. JClin Microbiol 6:445– 449.

54. Jacobson ES, Compton GM. 1996. Discordant regulation of phenoloxi-dase and capsular polysaccharide in Cryptococcus neoformans. J Med VetMycol 34:289 –291. http://dx.doi.org/10.1080/02681219680000491.

55. Missall TA, Moran JM, Corbett JA, Lodge JK. 2005. Distinct stressresponses of two functional laccases in Cryptococcus neoformans are re-vealed in the absence of the thiol-specific antioxidant Tsa1. Eukaryot Cell4:202–208. http://dx.doi.org/10.1128/EC.4.1.202-208.2005.

56. Pukkila-Worley R, Gerrald QD, Kraus PR, Boily MJ, Davis MJ, GilesSS, Cox GM, Heitman J, Alspaugh JA. 2005. Transcriptional network ofmultiple capsule and melanin genes governed by the Cryptococcus neo-formans cyclic AMP cascade. Eukaryot Cell 4:190 –201. http://dx.doi.org/10.1128/EC.4.1.190-201.2005.

57. Zhu X, Williamson PR. 2004. Role of laccase in the biology and viru-lence of Cryptococcus neoformans. FEMS Yeast Res 5:1–10. http://dx.doi.org/10.1016/j.femsyr.2004.04.004.

58. Zhu XD, Gibbons J, Garcia-Rivera J, Casadevall A, Williamson PR.2001. Laccase of Cryptococcus neoformans is a cell wall-associated viru-lence factor. Infect Immun 69:5589 –5596. http://dx.doi.org/10.1128/IAI.69.9.5589-5596.2001.

59. Salas SD, Bennett JE, Kwon-Chung KJ, Perfect JR, Williamson PR.1996. Effect of the laccase gene CNLAC1, on virulence of Cryptococcusneoformans. J Exp Med 184:377–386. http://dx.doi.org/10.1084/jem.184.2.377.

60. Wang Y, Casadevall A. 1994. Susceptibility of melanized and non-melanized Cryptococcus neoformans to nitrogen- and oxygen-derived ox-idants. Infect Immun 62:3004 –3007.

61. Wang Y, Aisen P, Casadevall A. 1995. Cryptococcus neoformans melaninand virulence: mechanism of action. Infect Immun 63:3131–3136.

62. Jacobson ES, Tinnell SB. 1993. Antioxidant function of fungal melanin.J Bacteriol 175:7102–7104.

63. van Duin D, Casadevall A, Nosanchuk JD. 2002. Melanization ofCryptococcus neoformans and Histoplasma capsulatum reduces their sus-ceptibilities to amphotericin B and caspofungin. Antimicrob AgentsChemother 46:3394 –3400. http://dx.doi.org/10.1128/AAC.46.11.3394-3400.2002.

64. Ikeda R, Sugita T, Jacobson ES, Shinoda T. 2003. Effects of melaninupon susceptibility of Cryptococcus to antifungals. Microbiol Immunol47:271–277. http://dx.doi.org/10.1111/j.1348-0421.2003.tb03395.x.

65. Wang YL, Casadevall A. 1994. Growth of Cryptococcus neoformans inpresence of L-dopa decreases its susceptibility to amphotericin B. Anti-microb Agents Chemother 38:2648 –2650. http://dx.doi.org/10.1128/AAC.38.11.2648.

66. Almeida FB, Cerqueira FM, Silva Rdo N, Ulhoa CJ, Lima AL. 2007.Mycoparasitism studies of Trichoderma harzianum strains against Rhi-zoctonia solani: evaluation of coiling and hydrolytic enzyme production.Biotechnol Lett 29:1189 –1193. http://dx.doi.org/10.1007/s10529-007-9372-z.

67. Dos Reis Almeida FB, de Oliveira LL, de Sousa MV, Barreira MCR,Hanna ES. 2010. Paracoccin from Paracoccidioides brasiliensis; purifica-tion through affinity with chitin and identification of N-acetyl-beta-D-glucosaminidase activity. Yeast 27:67–76.

68. Dos Reis Almeida FB, Carvalho FC, Mariano VS, Alegre ACP, SilvaRD, Hanna ES, Roque-Barreira MC. 2011. Influence of N-glycosylationon the morphogenesis and growth of Paracoccidioides brasiliensis and onthe biological activities of yeast proteins. PLoS One 6:e29216. http://dx.doi.org/10.1371/journal.pone.0029216.

69. Dos Reis Almeida FB, Pigosso LL, de Lima Damasio AR, MonteiroVN, de Almeida Soares CM, Silva RN, Roque-Barreira MC. 2014.alpha-(1,4)-Amylase, but not alpha- and beta-(1,3)-glucanases, may beresponsible for the impaired growth and morphogenesis of Paracoccid-ioides brasiliensis induced by N-glycosylation inhibition. Yeast 31:1–11.http://dx.doi.org/10.1002/yea.2983.

70. Partridge BM, Winner HI. 1965. Cryptococcus neoformans in bird drop-pings in London. Lancet i:1060 –1061.

71. Walter JE, Yee RB. 1968. Factors that determine the growth of Crypto-coccus neoformans in avian excreta. Am J Epidemiol 88:445– 450.

72. Kwon-Chung KJ, Wickes BL, Booth JL, Vishniac HS, Bennett JE. 1987.Urease inhibition by EDTA in the two varieties of Cryptococcus neofor-mans. Infect Immun 55:1751–1754.

73. Vogel RA. 1969. Primary isolation medium for Cryptococcus neoformans.Appl Microbiol 18:1100.

74. Zimmer BL, Roberts GD. 1979. Rapid selective urease test for presump-tive identification of Cryptococcus neoformans. J Clin Microbiol 10:380 –381.

75. Cox GM, Mukherjee J, Cole GT, Casadevall A, Perfect JR. 2000. Ureaseas a virulence factor in experimental cryptococcosis. Infect Immun 68:443– 448. http://dx.doi.org/10.1128/IAI.68.2.443-448.2000.

76. Olszewski MA, Noverr MC, Chen GH, Toews GB, Cox GM, Perfect JR,Huffnagle GB. 2004. Urease expression by Cryptococcus neoformans pro-motes microvascular sequestration, thereby enhancing central nervoussystem invasion. Am J Pathol 164:1761–1771. http://dx.doi.org/10.1016/S0002-9440(10)63734-0.

77. Bava AJ, Negroni R, Bianchi M. 1993. Cryptococcosis produced by aurease negative strain of Cryptococcus neoformans. J Med Vet Mycol 31:87– 89. http://dx.doi.org/10.1080/02681219380000091.

78. Ruane PJ, Walker LJ, George WL. 1988. Disseminated infection causedby urease-negative Cryptococcus neoformans. J Clin Microbiol 26:2224 –2225.

79. Cazin J, Jr, Kozel TR, Lupan DM, Burt WR. 1969. Extracellulardeoxyribonuclease production by yeasts. J Bacteriol 100:760 –762.

80. Rocha JD, Nascimento MT, Decote-Ricardo D, Corte-Real S, MorrotA, Heise N, Nunes MP, Previato JO, Mendonca-Previato L, DosReisGA, Saraiva EM, Freire-de-Lima CG. 2015. Capsular polysaccharidesfrom Cryptococcus neoformans modulate production of neutrophil extra-cellular traps (NETs) by human neutrophils. Sci Rep 5:8008. http://dx.doi.org/10.1038/srep08008.

81. Sanchez M, Colom F. 2010. Extracellular DNase activity of Cryptococcusneoformans and Cryptococcus gattii. Rev Iberoam Micol 27:10 –13. http://dx.doi.org/10.1016/j.riam.2009.11.004.

82. Fridovich I. 1995. Superoxide radical and superoxide dismutases.Annu Rev Biochem 64:97–112. http://dx.doi.org/10.1146/annurev.bi.64.070195.000525.

83. Hamilton AJ, Holdom MD. 1997. Biochemical comparison of theCu,Zn superoxide dismutases of Cryptococcus neoformans var. neofor-mans and Cryptococcus neoformans var. gattii. Infect Immun 65:488 –494.

84. Tesfa-Selase F, Hay RJ. 1995. Superoxide dismutase of Cryptococcusneoformans: purification and characterization. J Med Vet Mycol 33:253–259. http://dx.doi.org/10.1080/02681219580000511.

85. Giles SS, Batinic-Haberle I, Perfect JR, Cox GM. 2005. Cryptococcusneoformans mitochondrial superoxide dismutase: an essential link be-tween antioxidant function and high-temperature growth. Eukaryot Cell4:46 –54. http://dx.doi.org/10.1128/EC.4.1.46-54.2005.

86. Narasipura SD, Ault JG, Behr MJ, Chaturvedi V, Chaturvedi S. 2003.Characterization of Cu,Zn superoxide dismutase (SOD1) gene knock-out mutant of Cryptococcus neoformans var. gattii: role in biology andvirulence. Mol Microbiol 47:1681–1694.

87. Narasipura SD, Chaturvedi V, Chaturvedi S. 2005. Characterization ofCryptococcus neoformans variety gattii SOD2 reveals distinct roles of thetwo superoxide dismutases in fungal biology and virulence. Mol Micro-biol 55:1782–1800. http://dx.doi.org/10.1111/j.1365-2958.2005.04503.x.

88. Siafakas AR, Wright LC, Sorrell TC, Djordjevic JT. 2006. Lipid rafts inCryptococcus neoformans concentrate the virulence determinants phos-pholipase B1 and Cu/Zn superoxide dismutase. Eukaryot Cell 5:488 –498. http://dx.doi.org/10.1128/EC.5.3.488-498.2006.

89. Cox GM, Harrison TS, McDade HC, Taborda CP, Heinrich G, Casa-devall A, Perfect JR. 2003. Superoxide dismutase influences the viru-lence of Cryptococcus neoformans by affecting growth within macro-

Minireview

December 2015 Volume 14 Number 12 ec.asm.org 1181Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 10: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

phages. Infect Immun 71:173–180. http://dx.doi.org/10.1128/IAI.71.1.173-180.2003.

90. Jacobson ES, Jenkins ND, Todd JM. 1994. Relationship between super-oxide-dismutase and melanin in a pathogenic fungus. Infect Immun62:4085– 4086.

91. Chen SC, Muller M, Zhou JZ, Wright LC, Sorrell TC. 1997. Phospho-lipase activity in Cryptococcus neoformans: a new virulence factor? J InfectDis 175:414 – 420. http://dx.doi.org/10.1093/infdis/175.2.414.

92. Chen SCA, Wright LC, Santangelo RT, Muller M, Moran VR, KuchelPW, Sorrell TC. 1997. Identification of extracellular phospholipase B,lysophospholipase, and acyltransferase produced by Cryptococcus neofor-mans. Infect Immun 65:405– 411.

93. Henry J, Guillotte A, Luberto C, Del Poeta M. 2011. Characterizationof inositol phospho-sphingolipid-phospholipase C 1 (Isc1) in Cryptococ-cus neoformans reveals unique biochemical features. FEBS Lett 585:635–640. http://dx.doi.org/10.1016/j.febslet.2011.01.015.

94. Barrett-Bee K, Hayes Y, Wilson RG, Ryley JF. 1985. A comparison ofphospholipase activity, cellular adherence and pathogenicity of yeasts. JGen Microbiol 131:1217–1221.

95. Santangelo R, Zoellner H, Sorrell T, Wilson C, Donald C, DjordjevicJ, Shounan Y, Wright L. 2004. Role of extracellular phospholipases andmononuclear phagocytes in dissemination of cryptococcosis in a murinemodel. Infect Immun 72:2229 –2239. http://dx.doi.org/10.1128/IAI.72.4.2229-2239.2004.

96. Chen SC, Wright LC, Golding JC, Sorrell TC. 2000. Purification andcharacterization of secretory phospholipase B, lysophospholipase andlysophospholipase/transacylase from a virulent strain of the pathogenicfungus Cryptococcus neoformans. Biochem J 347:431– 439. http://dx.doi.org/10.1042/bj3470431.

97. Siafakas AR, Sorrell TC, Wright LC, Wilson C, Larsen M, Boadle R,Williamson PR, Djordjevic JT. 2007. Cell wall-linked cryptococcalphospholipase B1 is a source of secreted enzyme and a determinant of cellwall integrity. J Biol Chem 282:37508 –37514. http://dx.doi.org/10.1074/jbc.M707913200.

98. Wright LC, Santangelo RM, Ganendren R, Payne J, Djordjevic JT,Sorrell TC. 2007. Cryptococcal lipid metabolism: phospholipase B1 isimplicated in transcellular metabolism of macrophage-derived lipids.Eukaryot Cell 6:37– 47. http://dx.doi.org/10.1128/EC.00262-06.

99. Noverr MC, Cox GM, Perfect JR, Huffnagle GB. 2003. Role of PLB1 inpulmonary inflammation and cryptococcal eicosanoid production. In-fect Immun 71:1538 –1547. http://dx.doi.org/10.1128/IAI.71.3.1538-1547.2003.

100. Turner KM, Wright LC, Sorrell TC, Djordjevic JT. 2006. N-linkedglycosylation sites affect secretion of cryptococcal phospholipase B1, ir-respective of glycosylphosphatidylinositol anchoring. Biochim BiophysActa 1760:1569 –1579. http://dx.doi.org/10.1016/j.bbagen.2006.07.002.

101. Chayakulkeeree M, Sorrell TC, Siafakas AR, Wilson CF, Pantarat N,Gerik KJ, Boadle R, Djordjevic JT. 2008. Role and mechanism of phos-phatidylinositol-specific phospholipase C in survival and virulence ofCryptococcus neoformans. Mol Microbiol 69:809 – 826. http://dx.doi.org/10.1111/j.1365-2958.2008.06310.x.

102. Lev S, Desmarini D, Li C, Chayakulkeeree M, Traven A, Sorrell TC,Djordjevic JT. 2013. Phospholipase C of Cryptococcus neoformans regu-lates homeostasis and virulence by providing inositol trisphosphate as asubstrate for Arg1 kinase. Infect Immun 81:1245–1255. http://dx.doi.org/10.1128/IAI.01421-12.

103. Bauman AL, Scott JD. 2002. Kinase- and phosphatase-anchoring pro-teins: harnessing the dynamic duo. Nat Cell Biol 4:E203–E206. http://dx.doi.org/10.1038/ncb0802-e203.

104. McConnell JL, Wadzinski BE. 2009. Targeting protein serine/threoninephosphatases for drug development. Mol Pharmacol 75:1249 –1261.http://dx.doi.org/10.1124/mol.108.053140.

105. Collopy-Junior I, Esteves FF, Nimrichter L, Rodrigues ML, AlvianoCS, Meyer-Fernandes JR. 2006. An ectophosphatase activity in Crypto-coccus neoformans. FEMS Yeast Res 6:1010 –1017. http://dx.doi.org/10.1111/j.1567-1364.2006.00105.x.

106. Lev S, Crossett B, Cha SY, Desmarini D, Li C, Chayakulkeeree M,Wilson CF, Williamson PR, Sorrell TC, Djordjevic JT. 2014. Identifi-cation of Aph1, a phosphate-regulated, secreted, and vacuolar acid phos-phatase in Cryptococcus neoformans. mBio 5:e01649-14. http://dx.doi.org/10.1128/mBio.01649-14.

107. Brueske CH. 1986. Proteolytic activity of a clinical isolate of Cryptococcusneoformans. J Clin Microbiol 23:631– 633.

108. Chen LC, Blank ES, Casadevall A. 1996. Extracellular proteinase activityof Cryptococcus neoformans. Clin Diagn Lab Immunol 3:570 –574.

109. Ruma-Haynes P, Brownlee AG, Sorrell TC. 2000. A rapid method fordetecting extracellular proteinase activity in Cryptococcus neoformansand a survey of 63 isolates. J Med Microbiol 49:733–737. http://dx.doi.org/10.1099/0022-1317-49-8-733.

110. Il Yoo J, Lee YS, Song CY, Kim BS. 2004. Purification and character-ization of a 43-kilodalton extracellular serine proteinase from Cryptococ-cus neoformans. J Clin Microbiol 42:722–726. http://dx.doi.org/10.1128/JCM.42.2.722-726.2004.

111. Pinti M, Orsi CF, Gibellini L, Esposito R, Cossarizza A, Blasi E,Peppoloni S, Mussini C. 2007. Identification and characterization of anaspartyl protease from Cryptococcus neoformans. FEBS Lett 581:3882–3886. http://dx.doi.org/10.1016/j.febslet.2007.07.006.

112. Chen LC, Pirofski LA, Casadevall A. 1997. Extracellular proteins ofCryptococcus neoformans and host antibody response. Infect Immun 65:2599 –2605.

113. Vu K, Tham R, Uhrig JP, Thompson GR, III, Na Pombejra S, Jam-klang M, Bautos JM, Gelli A. 2014. Invasion of the central nervoussystem by Cryptococcus neoformans requires a secreted fungal metallo-protease. mBio 5:e01101-14. http://dx.doi.org/10.1128/mBio.01101-14.

114. Perfect JR, Toffaletti DL, Rude TH. 1993. The gene encoding phospho-ribosylaminoimidazole carboxylase (Ade2) is essential for growth ofCryptococcus neoformans in cerebrospinal fluid. Infect Immun 61:4446 –4451.

115. Garrad RC, Bhattacharjee JK. 1992. Lysine biosynthesis in selectedpathogenic fungi— characterization of lysine auxotrophs and the clonedLys1 gene of Candida albicans. J Bacteriol 174:7379 –7384.

116. Lodge JK, Johnson RL, Weinberg RA, Gordon JI. 1994. Comparison ofmyristoyl-CoA:protein N-myristoyltransferases from three pathogenicfungi: Cryptococcus neoformans, Histoplasma capsulatum, and Candidaalbicans. J Biol Chem 269:2996 –3009.

117. Lodge JK, Jackson-Machelski E, Toffaletti DL, Perfect JR, Gordon JI.1994. Targeted gene replacement demonstrates that myristoyl-CoA/protein N-myristoyltransferase is essential for viability of Cryptococcusneoformans. Proc Natl Acad Sci U S A 91:12008 –12012. http://dx.doi.org/10.1073/pnas.91.25.12008.

118. Langner CA, Lodge JK, Travis SJ, Caldwell JE, Lu TB, Li Q, BryantML, Devadas B, Gokel GW, Kobayashi GS, Gordon JI. 1992. 4-Oxa-tetradecanoic acid is fungicidal for Cryptococcus neoformans and inhibitsreplication of human immunodeficiency virus I. J Biol Chem 267:17159 –17169.

119. Lamb DC, Corran A, Baldwin BC, Kwon-Chung J, Kelly SL. 1995.Resistant P45051A1 activity in azole antifungal tolerant Cryptococcusneoformans from AIDS patients. FEBS Lett 368:326 –330. http://dx.doi.org/10.1016/0014-5793(95)00684-2.

120. Bozzette SA, Larsen RA, Chiu J, Leal MAE, Jacobsen J, Rothman P,Robinson P, Gilbert G, Mccutchan JA, Tilles J, Leedom JM, RichmanDD. 1991. A placebo-controlled trial of maintenance therapy with flu-conazole after treatment of cryptococcal meningitis in the acquired im-munodeficiency syndrome. N Engl J Med 324:580 –584. http://dx.doi.org/10.1056/NEJM199102283240902.

121. Anonymous. 1980. Garlic in cryptococcal meningitis: a preliminary re-port of 21 cases. Chin Med J (Engl) 93:123–126.

122. Haynes MP, Chong PLG, Buckley HR, Pieringer RA. 1996. Fluores-cence studies on the molecular action of amphotericin B on susceptibleand resistant fungal cells. Biochemistry 35:7983–7992. http://dx.doi.org/10.1021/bi952910c.

123. Venkateswarlu K, Taylor M, Manning NJ, Rinaldi MG, Kelly SL. 1997.Fluconazole tolerance in clinical isolates of Cryptococcus neoformans. An-timicrob Agents Chemother 41:748 –751.

124. Kelly SL, Lamb DC, Taylor M, Corran AJ, Baldwin BC, Powderly WG.1994. Resistance to amphotericin B associated with defective sterol delta8¡7 isomerase in a Cryptococcus neoformans strain from an AIDS pa-tient. FEMS Microbiol Lett 122:39 – 42. http://dx.doi.org/10.1111/j.1574-6968.1994.tb07140.x.

125. Luberto C, Toffaletti DL, Wills EA, Tucker SC, Casadevall A, PerfectJR, Hannun YA, Del Poeta M. 2001. Roles for inositol-phosphorylceramide synthase 1 (IPC1) in pathogenesis of C. neoformans. Genes Dev15:201–212. http://dx.doi.org/10.1101/gad.856001.

126. Shea JM, Kechichian TB, Luberto C, Del Poeta M. 2006. The crypto-coccal enzyme inositol phosphosphingolipid-phospholipase C confersresistance to the antifungal effects of macrophages and promotes fungal

Minireview

1182 ec.asm.org December 2015 Volume 14 Number 12Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 11: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

dissemination to the central nervous system. Infect Immun 74:5977–5988. http://dx.doi.org/10.1128/IAI.00768-06.

127. Del Poeta M, Nimrichter L, Rodrigues ML, Luberto C. 2014. Synthesisand biological properties of fungal glucosylceramide. PLoS Pathog 10:e1003832. http://dx.doi.org/10.1371/journal.ppat.1003832.

128. Rittershaus PC, Kechichian TB, Allegood JC, Merrill AH, Hennig M,Luberto C, Del Poeta M. 2006. Glucosylceramide synthase is an essentialregulator of pathogenicity of Cryptococcus neoformans. J Clin Invest 116:1651–1659. http://dx.doi.org/10.1172/JCI27890.

129. Nosanchuk JD, Ovalle R, Casadevall A. 2001. Glyphosate inhibitsmelanization of Cryptococcus neoformans and prolongs survival of miceafter systemic infection. J Infect Dis 183:1093–1099. http://dx.doi.org/10.1086/319272.

130. De Clercq E. 2013. The nucleoside reverse transcriptase inhibitors, non-nucleoside reverse transcriptase inhibitors, and protease inhibitors in thetreatment of HIV infections (AIDS). Adv Pharmacol 67:317–358. http://dx.doi.org/10.1016/B978-0-12-405880-4.00009-3.

131. Loregian A, Mercorelli B, Nannetti G, Compagnin C, Palu G. 2014.Antiviral strategies against influenza virus: towards new therapeutic ap-proaches. Cell Mol Life Sci 71:3659 –3683. http://dx.doi.org/10.1007/s00018-014-1615-2.

132. Cassone A, De Bernardis F, Torosantucci A, Tacconelli E, TumbarelloM, Cauda R. 1999. In vitro and in vivo anticandidal activity of humanimmunodeficiency virus protease inhibitors. J Infect Dis 180:448 – 453.http://dx.doi.org/10.1086/314871.

133. Blasi E, Colombari B, Orsi CF, Pinti M, Troiano L, Cossarizza A,Esposito R, Peppoloni S, Mussini C, Neglia R. 2004. The humanimmunodeficiency virus (HIV) protease inhibitor indinavir directly af-fects the opportunistic fungal pathogen Cryptococcus neoformans. FEMSImmunol Med Microbiol 42:187–195. http://dx.doi.org/10.1016/j.femsim.2004.05.001.

134. Campbell LT, Chen C, Ferdous J, Padula MP, Harry E, Hofer M,Campbell IL, Carter DA. 2015. Cryptococcus strains with differentpathogenic potential have diverse protein secretomes. Eukaryot Cell 14:554 –563. http://dx.doi.org/10.1128/EC.00052-15.

135. Maligie MA, Selitrennikoff CP. 2005. Cryptococcus neoformans resis-tance to echinocandins: (1,3)beta-glucan synthase activity is sensitive toechinocandins. Antimicrob Agents Chemother 49:2851–2856. http://dx.doi.org/10.1128/AAC.49.7.2851-2856.2005.

136. Casal M, Linares MJ. 1983. Contribution to the study of the enzymaticprofiles of yeast organisms with medical interest. Mycopathologia 81:155–159. http://dx.doi.org/10.1007/BF00436820.

137. Mason DL, Wilson CL. 1979. Cytochemical and biochemical identifi-cation of lysosomes in Cryptococcus neoformans. Mycopathologia 68:183–190. http://dx.doi.org/10.1007/BF00578528.

138. Janbon G, Himmelreich U, Moyrand F, Improvisi L, Dromer F. 2001.Cas1p is a membrane protein necessary for the O-acetylation of the Cryp-tococcus neoformans capsular polysaccharide. Mol Microbiol 42:453–467. http://dx.doi.org/10.1046/j.1365-2958.2001.02651.x.

139. Baker LG, Specht CA, Donlin MJ, Lodge JK. 2007. Chitosan, thedeacetylated form of chitin, is necessary for cell wall integrity in Crypto-coccus neoformans. Eukaryot Cell 6:855– 867. http://dx.doi.org/10.1128/EC.00399-06.

140. Banks IR, Specht CA, Donlin MJ, Gerik KJ, Levitz SM, Lodge JK. 2005.A chitin synthase and its regulator protein are critical for chitosan pro-duction and growth of the fungal pathogen Cryptococcus neoformans.Eukaryot Cell 4:1902–1912. http://dx.doi.org/10.1128/EC.4.11.1902-1912.2005.

141. Baker LG, Specht CA, Lodge JK. 2009. Chitinases are essential for sexualdevelopment but not vegetative growth in Cryptococcus neoformans. Eu-karyot Cell 8:1692–1705. http://dx.doi.org/10.1128/EC.00227-09.

142. Polacheck I, Kwon-Chung KJ. 1980. Creatinine metabolism in Crypto-coccus neoformans and Cryptococcus bacillisporus. J Bacteriol 142:15–20.

143. Wills EA, Roberts IS, Del Poeta M, Rivera J, Casadevall A, Cox GM,Perfect JR. 2001. Identification and characterization of the Cryptococcusneoformans phosphomannose isomerase-encoding gene, MAN1, and itsimpact on pathogenicity. Mol Microbiol 40:610 – 620. http://dx.doi.org/10.1046/j.1365-2958.2001.02401.x.

144. Cottrell TR, Griffith CL, Liu H, Nenninger AA, Doering TL. 2007. Thepathogenic fungus Cryptococcus neoformans expresses two functionalGDP-mannose transporters with distinct expression patterns and rolesin capsule synthesis. Eukaryot Cell 6:776 –785. http://dx.doi.org/10.1128/EC.00015-07.

145. Biondo C, Beninati C, Bombaci M, Messina L, Mancuso G, MidiriA, Galbo R, Teti G. 2003. Induction of T helper type 1 responses bya polysaccharide deacetylase from Cryptococcus neoformans. Infect Im-mun 71:5412–5417. http://dx.doi.org/10.1128/IAI.71.9.5412-5417.2003.

146. Erickson T, Liu L, Gueyikian A, Zhu XD, Gibbons J, Williamson PR.2001. Multiple virulence factors of Cryptococcus neoformans are depen-dent on VPH1. Mol Microbiol 42:1121–1131. http://dx.doi.org/10.1046/j.1365-2958.2001.02712.x.

147. Bar-Peled M, Griffith CL, Doering TL. 2001. Functional cloning andcharacterization of a UDP-glucuronic acid decarboxylase: the patho-genic fungus Cryptococcus neoformans elucidates UDP-xylose synthesis.Proc Natl Acad Sci U S A 98:12003–12008. http://dx.doi.org/10.1073/pnas.211229198.

148. Levitz SM, Specht CA. 2006. The molecular basis for the immunogenic-ity of Cryptococcus neoformans mannoproteins. FEMS Yeast Res 6:513–524. http://dx.doi.org/10.1111/j.1567-1364.2006.00071.x.

149. White CW, Jacobson ES. 1993. Mannosyl transfer in Cryptococcus neo-formans. Can J Microbiol 39:129 –133. http://dx.doi.org/10.1139/m93-019.

150. Fiskin AM, Zalles MC, Garrison RG. 1990. Electron cytochemicalstudies of Cryptococcus neoformans grown on uric acid and relatedsources of nitrogen. J Med Vet Mycol 28:197–207. http://dx.doi.org/10.1080/02681219080000261.

151. Biondo C, Mancuso G, Midiri A, Bombaci M, Messina L, Beninati C,Teti G. 2006. Identification of major proteins secreted by Cryptococcusneoformans. FEMS Yeast Res 6:645– 651. http://dx.doi.org/10.1111/j.1567-1364.2006.00043.x.

152. Eigenheer RA, Jin Lee Y, Blumwald E, Phinney BS, Gelli A. 2007.Extracellular glycosylphosphatidylinositol-anchored mannoproteinsand proteases of Cryptococcus neoformans. FEMS Yeast Res 7:499 –510.http://dx.doi.org/10.1111/j.1567-1364.2006.00198.x.

153. Bien CM, Chang YC, Nes WD, Kwon-Chung KJ, Espenshade PJ. 2009.Cryptococcus neoformans site-2 protease is required for virulence andsurvival in the presence of azole drugs. Mol Microbiol 74:672– 690. http://dx.doi.org/10.1111/j.1365-2958.2009.06895.x.

154. Miyakoshi S, Uchiyama H, Someya T, Satoh T, Tabuchi T. 1987.Distribution of the methylcitric acid cycle and beta-oxidation pathwayfor propionate catabolism in fungi. Agric Biol Chem 51:2381–2387. http://dx.doi.org/10.1271/bbb1961.51.2381.

155. Vanden Bossche H, Marichal P, Le Jeune L, Coene MC, Gorrens J,Cools W. 1993. Effects of itraconazole on cytochrome P-450-dependentsterol 14 alpha-demethylation and reduction of 3-ketosteroids in Cryp-tococcus neoformans. Antimicrob Agents Chemother 37:2101–2105. http://dx.doi.org/10.1128/AAC.37.10.2101.

156. Niehaus WG, Richardson SB, Wolz RL. 1996. Slow-binding inhibitionof 6-phosphogluconate dehydrogenase by zinc ion. Arch Biochem Bio-phys 333:333–337. http://dx.doi.org/10.1006/abbi.1996.0399.

157. Niehaus WG, White RH, Richardson SB, Bourne A, Ray WK. 1995.Polyethylene sulfonate: a tight-binding inhibitor of 6-phosphogluconatedehydrogenase of Cryptococcus neoformans. Arch Biochem Biophys 324:325–330. http://dx.doi.org/10.1006/abbi.1995.0045.

158. Thaker TM, Tanabe M, Fowler ML, Preininger AM, Ingram-Smith C,Smith KS, Iverson TM. 2013. Crystal structures of acetate kinases fromthe eukaryotic pathogens Entamoeba histolytica and Cryptococcus neofor-mans. J Struct Biol 181:185–189. http://dx.doi.org/10.1016/j.jsb.2012.11.001.

159. Missall TA, Pusateri ME, Donlin MJ, Chambers KT, Corbett JA, LodgeJK. 2006. Posttranslational, translational, and transcriptional responsesto nitric oxide stress in Cryptococcus neoformans: implications for viru-lence. Eukaryot Cell 5:518 –529. http://dx.doi.org/10.1128/EC.5.3.518-529.2006.

160. Alspaugh JA, Pukkila-Worley R, Harashima T, Cavallo LM, Funnell D,Cox GM, Perfect JR, Kronstad JW, Heitman J. 2002. Adenylyl cyclasefunctions downstream of the G alpha protein Gpa1 and controls matingand pathogenicity of Cryptococcus neoformans. Eukaryot Cell 1:75– 84.http://dx.doi.org/10.1128/EC.1.1.75-84.2002.

161. Akhter S, McDade HC, Gorlach JM, Heinrich G, Cox GM, Perfect JR.2003. Role of alternative oxidase gene in pathogenesis of Cryptococcusneoformans. Infect Immun 71:5794 –5802. http://dx.doi.org/10.1128/IAI.71.10.5794-5802.2003.

162. Bahn YS, Cox GM, Perfect JR, Heitman J. 2005. Carbonic anhydraseand CO2 sensing during Cryptococcus neoformans growth, differentia-

Minireview

December 2015 Volume 14 Number 12 ec.asm.org 1183Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 12: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

tion, and virulence. Curr Biol 15:2013–2020. http://dx.doi.org/10.1016/j.cub.2005.09.047.

163. Mogensen EG, Janbon G, Chaloupka J, Steegborn C, Fu MS, MoyrandF, Klengel T, Pearson DS, Geeves MA, Buck J, Levin LR, MuhlschlegelFA. 2006. Cryptococcus neoformans senses CO2 through the carbonicanhydrase Can2 and the adenylyl cyclase Cac1. Eukaryot Cell 5:103–111.http://dx.doi.org/10.1128/EC.5.1.103-111.2006.

164. Wang YN, Liu TB, Patel S, Jiang LH, Xue CY. 2011. The casein kinaseI protein Cck1 regulates multiple signaling pathways and is essential forcell integrity and fungal virulence in Cryptococcus neoformans. EukaryotCell 10:1455–1464. http://dx.doi.org/10.1128/EC.05207-11.

165. Giles SS, Perfect JR, Cox GM. 2005. Cytochrome c peroxidase contrib-utes to the antioxidant defense of Cryptococcus neoformans. Fungal GenetBiol 42:20 –29. http://dx.doi.org/10.1016/j.fgb.2004.09.003.

166. Biondo C, Beninati C, Delfino D, Oggioni M, Mancuso G, Midiri A,Bombaci M, Tomaselli G, Teti G. 2002. Identification and cloning of acryptococcal deacetylase that produces protective immune responses.Infect Immun 70:2383–2391. http://dx.doi.org/10.1128/IAI.70.5.2383-2391.2002.

167. Kelleher DJ, Banerjee S, Cura AJ, Samuelson J, Gilmore R. 2007.Dolichol-linked oligosaccharide selection by the oligosaccharyltrans-ferase in protist and fungal organisms. J Cell Biol 177:29 –37. http://dx.doi.org/10.1083/jcb.200611079.

168. Idnurm A, Heitman J. 2010. Ferrochelatase is a conserved downstreamtarget of the blue light-sensing white collar complex in fungi. Microbiol-ogy 156:2393–2407. http://dx.doi.org/10.1099/mic.0.039222-0.

169. Hu GG, Kronstad JW. 2010. A putative P-type ATPase, Apt1, is involvedin stress tolerance and virulence in Cryptococcus neoformans. EukaryotCell 9:74 – 83. http://dx.doi.org/10.1128/EC.00289-09.

170. Rizzo J, Oliveira DL, Joffe LS, Hu G, Gazos-Lopes F, Fonseca FL,Almeida IC, Frases S, Kronstad JW, Rodrigues ML. 2014. Role of theApt1 protein in polysaccharide secretion by Cryptococcus neoformans.Eukaryot Cell 13:715–726. http://dx.doi.org/10.1128/EC.00273-13.

171. Niehaus WG, Mallett TC. 1994. Purification and characterization ofglucose-6-phosphate dehydrogenase from Cryptococcus neoformansidentification as “nothing dehydrogenase.” Arch Biochem Biophys 313:304 –309.

172. Safrin RE, Lancaster LA, Davis CE, Braude AI. 1986. Differentiation ofCryptococcus neoformans serotypes by isoenzyme electrophoresis. Am JClin Pathol 86:204 –208.

173. Missall TA, Cherry-Harris JF, Lodge JK. 2005. Two glutathione per-oxidases in the fungal pathogen Cryptococcus neoformans are expressed inthe presence of specific substrates. Microbiology 151:2573–2581. http://dx.doi.org/10.1099/mic.0.28132-0.

174. Kingsbury JM, McCusker JH. 2008. Threonine biosynthetic genes areessential in Cryptococcus neoformans. Microbiology 154:2767–2775. http://dx.doi.org/10.1099/mic.0.2008/019729-0.

175. Nazi I, Scott A, Sham A, Rossi L, Williamson PR, Kronstad JW,Wright GD. 2007. Role of homoserine transacetylase as a new target forantifungal agents. Antimicrob Agents Chemother 51:1731–1736. http://dx.doi.org/10.1128/AAC.01400-06.

176. Jong A, Wu CH, Chen HM, Luo F, Kwon-Chung KJ, Chang YC,LaMunyon CW, Plaas A, Huang SH. 2007. Identification and charac-terization of CPS1 as a hyaluronic acid synthase contributing to thepathogenesis of Cryptococcus neoformans infection. Eukaryot Cell6:1486 –1496. http://dx.doi.org/10.1128/EC.00120-07.

177. Parker AR, Moore TD, Edman JC, Schwab JM, Davisson VJ. 1994.Cloning, sequence analysis and expression of the gene encoding imida-zole glycerol phosphate dehydratase in Cryptococcus neoformans. Gene145:135–138. http://dx.doi.org/10.1016/0378-1119(94)90336-0.

178. Morrow CA, Stamp A, Valkov E, Kobe B, Fraser JA. 2010. Crystal-lization and preliminary X-ray analysis of mycophenolic acid-resistant and mycophenolic acid-sensitive forms of IMP dehydroge-nase from the human fungal pathogen Cryptococcus. Acta CrystallogrSect F Struct Biol Cryst Commun 66:1104 –1107. http://dx.doi.org/10.1107/S1744309110031659.

179. Heung LJ, Luberto C, Plowden A, Hannun YA, Del Poeta M. 2004. Thesphingolipid pathway regulates Pkc1 through the formation of diacyl-glycerol in Cryptococcus neoformans. J Biol Chem 279:21144 –21153.http://dx.doi.org/10.1074/jbc.M312995200.

180. Cheon SA, Jung KW, Chen YL, Heitman J, Bahn YS, Kang HA.2011. Unique evolution of the UPR pathway with a novel bZIP tran-scription factor, Hxl1, for controlling pathogenicity of Cryptococcus

neoformans. PLoS Pathog 7:e1002177. http://dx.doi.org/10.1371/journal.ppat.1002177.

181. Rude TH, Toffaletti DL, Cox GM, Perfect JR. 2002. Relationship of theglyoxylate pathway to the pathogenesis of Cryptococcus neoformans. In-fect Immun 70:5684 –5694. http://dx.doi.org/10.1128/IAI.70.10.5684-5694.2002.

182. Mahmoud YA, el Souod SM, Niehaus WG. 1995. Purification andcharacterization of malate dehydrogenase from Cryptococcus neofor-mans. Arch Biochem Biophys 322:69 –75. http://dx.doi.org/10.1006/abbi.1995.1437.

183. Perfect JR, Rude TH, Wong B, Flynn T, Chaturvedi V, Niehaus W.1996. Identification of a Cryptococcus neoformans gene that directs ex-pression of the cryptic Saccharomyces cerevisiae mannitol dehydrogenasegene. J Bacteriol 178:5257–5262.

184. Suvarna K, Bartiss A, Wong B. 2000. Mannitol-1-phosphate dehydro-genase from Cryptococcus neoformans is a zinc-containing long-chainalcohol/polyol dehydrogenase. Microbiology 146:2705–2713. http://dx.doi.org/10.1099/00221287-146-10-2705.

185. Olson GM, Fox DS, Wang P, Alspaugh JA, Buchanan KL. 2007. Roleof protein O-mannosyltransferase Pmt4 in the morphogenesis and viru-lence of Cryptococcus neoformans. Eukaryot Cell 6:222–234. http://dx.doi.org/10.1128/EC.00182-06.

186. Hicks JK, Bahn YS, Heitman J. 2005. Pde1 phosphodiesterase modu-lates cyclic AMP levels through a protein kinase A-mediated negativefeedback loop in Cryptococcus neoformans. Eukaryot Cell 4:1971–1981.http://dx.doi.org/10.1128/EC.4.12.1971-1981.2005.

187. Hast MA, Nichols CB, Armstrong SM, Kelly SM, Hellinga HW,Alspaugh JA, Beese LS. 2011. Structures of Cryptococcus neoformansprotein farnesyltransferase reveal strategies for developing inhibitors thattarget fungal pathogens. J Biol Chem 286:35149 –35162. http://dx.doi.org/10.1074/jbc.M111.250506.

188. Gerik KJ, Donlin MJ, Soto CE, Banks AM, Banks IR, Maligie MA,Selitrennikoff CP, Lodge JK. 2005. Cell wall integrity is dependent onthe PKC1 signal transduction pathway in Cryptococcus neoformans.Mol Microbiol 58:393– 408. http://dx.doi.org/10.1111/j.1365-2958.2005.04843.x.

189. Jin LH, Kryukov K, Suzuki Y, Imanishi T, Ikeo K, Gojobori T. 2009.The evolutionary study of small RNA-directed gene silencing pathwaysby investigating RNase III enzymes. Gene 435:1– 8. http://dx.doi.org/10.1016/j.gene.2008.12.022.

190. Kingsbury JM, Yang Z, Ganous TM, Cox GM, McCusker JH. 2004.Novel chimeric spermidine synthase-saccharopine dehydrogenasegene (SPE3-LYS9) in the human pathogen Cryptococcus neoformans.Eukaryot Cell 3:752–763. http://dx.doi.org/10.1128/EC.3.3.752-763.2004.

191. Ternes P, Sperling P, Albrecht S, Franke S, Cregg JM, Warnecke D,Heinz E. 2006. Identification of fungal sphingolipid C9-methyltransferases by phylogenetic profiling. J Biol Chem 281:5582–5592.

192. Zhu J, Lu J, Zhou Y, Li Y, Cheng J, Zheng C. 2006. Design, synthesis,and antifungal activities in vitro of novel tetrahydroisoquinoline com-pounds based on the structure of lanosterol 14alpha-demethylase(CYP51) of fungi. Bioorg Med Chem Lett 16:5285–5289. http://dx.doi.org/10.1016/j.bmcl.2006.08.001.

193. Nes WD, Zhou W, Ganapathy K, Liu J, Vatsyayan R, Chamala S,Hernandez K, Miranda M. 2009. Sterol 24-C-methyltransferase: anenzymatic target for the disruption of ergosterol biosynthesis and ho-meostasis in Cryptococcus neoformans. Arch Biochem Biophys 481:210 –218. http://dx.doi.org/10.1016/j.abb.2008.11.003.

194. Missall TA, Pusateri ME, Lodge JK. 2004. Thiol peroxidase is critical forvirulence and resistance to nitric oxide and peroxide in the fungal patho-gen, Cryptococcus neoformans. Mol Microbiol 51:1447–1458. http://dx.doi.org/10.1111/j.1365-2958.2004.03921.x.

195. Missall TA, Lodge JK. 2005. Thioredoxin reductase is essential for via-bility in the fungal pathogen Cryptococcus neoformans. Eukaryot Cell4:487– 489. http://dx.doi.org/10.1128/EC.4.2.487-489.2005.

196. Livi LL, Edman U, Schneider GP, Greene PJ, Santi DV. 1994.Cloning, expression and characterization of thymidylate synthasefrom Cryptococcus neoformans. Gene 150:221–226. http://dx.doi.org/10.1016/0378-1119(94)90430-8.

197. Petzold EW, Himmelreich U, Mylonakis E, Rude T, Toffaletti D, CoxGM, Miller JL, Perfect JR. 2006. Characterization and regulation of thetrehalose synthesis pathway and its importance in the pathogenicity of

Minireview

1184 ec.asm.org December 2015 Volume 14 Number 12Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from

Page 13: Virulence-Associated Enzymes of Cryptococcus neoformans · TABLE 1 Described enzymes in Cryptococcus neoformans Enzyme Function(s)a EC no. Reference(s) Localized on capsule and/or

Cryptococcus neoformans. Infect Immun 74:5877–5887. http://dx.doi.org/10.1128/IAI.00624-06.

198. Beverley SM, Owens KL, Showalter M, Griffith CL, Doering TL, JonesVC, McNeil MR. 2005. Eukaryotic UDP-galactopyranose mutase (GLFgene) in microbial and metazoal pathogens. Eukaryot Cell 4:1147–1154.http://dx.doi.org/10.1128/EC.4.6.1147-1154.2005.

199. Jacobson ES. 1987. Cryptococcal UDP-glucose dehydrogenase: enzymiccontrol of capsular biosynthesis. J Med Vet Mycol 25:131–135. http://dx.doi.org/10.1080/02681218780000201.

200. Reilly MC, Levery SB, Castle SA, Klutts JS, Doering TL. 2009. A novel

xylosylphosphotransferase activity discovered in Cryptococcus neofor-mans. J Biol Chem 284:36118 –36127. http://dx.doi.org/10.1074/jbc.M109.056226.

201. Block ER, Jennings AE, Bennett JE. 1973. 5-Fluorocytosine resistance inCryptococcus neoformans. Antimicrob Agents Chemother 3:649 – 656.http://dx.doi.org/10.1128/AAC.3.6.649.

202. Schwarz P, Janbon G, Dromer F, Lortholary O, Dannaoui E. 2007.Combination of amphotericin B with flucytosine is active in vitro againstflucytosine-resistant isolates of Cryptococcus neoformans. AntimicrobAgents Chemother 51:383–385. http://dx.doi.org/10.1128/AAC.00446-06.

Minireview

December 2015 Volume 14 Number 12 ec.asm.org 1185Eukaryotic Cell

on January 3, 2020 by guesthttp://ec.asm

.org/D

ownloaded from